1887

Abstract

Antimicrobial resistance (AMR) is of increasing global concern, threatening to undermine recent progress in reducing child and neonatal mortality. Repurposing older antimicrobials is a prominent strategy to combat multidrug-resistant sepsis. A potential agent is fosfomycin, however, there is scarce data regarding its activity and pharmacokinetics in the paediatric population.

We analysed a contemporary, systematically collected archive of community-acquired (CA) and hospital-acquired (HA) paediatric Gram-negative bacteraemia isolates for their susceptibility to fosfomcyin. MICs were determined using agar serial dilution methods and validated by disk diffusion testing where breakpoints are available. Disk diffusion antimicrobial susceptibility testing was also conducted for current empirical therapies (ampicillin, gentamicin, ceftriaxone) and amikacin (proposed in the literature as a new combination empirical therapeutic option).

Fosfomycin was highly active against invasive Gram-negative isolates, including 90  % (202/224) of and 96  % (22/23) of spp. Fosfomycin showed high sensitivity against both CA isolates (94 %, 142/151) and HA isolates (81 %, 78/96; =0.0015). CA isolates were significantly more likely to be susceptible to fosfomycin than the current first-line empirical therapy (96  % vs 59  %, <0.0001). Extended spectrum -lactamases (ESBL) production was detected in 34  % (85/247) of isolates with no significant difference in fosfomycin susceptibility between ESBL-positive or -negative isolates [73/85 (86  %) vs 147/162 (91  %) respectively, =0.245]. All isolates were susceptible to a fosfomycin-amikacin combination.

Gram-negative paediatric bacteraemia isolates are highly susceptible to fosfomycin, which could be combined with aminoglycosides as a new, carbapenem-sparing regimen to achieve excellent coverage to treat antimicrobial-resistant neonatal and paediatric sepsis.

  • This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
Loading

Article metrics loading...

/content/journal/jmm/10.1099/jmm.0.000973
2019-05-01
2024-04-23
Loading full text...

Full text loading...

/deliver/fulltext/jmm/68/5/711.html?itemId=/content/journal/jmm/10.1099/jmm.0.000973&mimeType=html&fmt=ahah

References

  1. Williams PCM, Isaacs D, Berkley JA. Antimicrobial resistance among children in sub-Saharan Africa. Lancet Infect Dis 2017
    [Google Scholar]
  2. Laxminarayan R, Matsoso P, Pant S et al. Access to effective antimicrobials: a worldwide challenge. Lancet 2016; 387:168–175
    [Google Scholar]
  3. The World Health Organization Pocket Book of Hospital Care for Children: Guidelines for the Management of Common Illnesses with Limited Resources 2013
    [Google Scholar]
  4. Downie L, Armiento R, Subhi R, Kelly J, Clifford V et al. Community-acquired neonatal and infant sepsis in developing countries: efficacy of WHO’s currently recommended antibiotics- systematic review and meta-analysis. Arch Dis Childhood 2013; 98:146–154
    [Google Scholar]
  5. Le Doare K, Bielicki J, Heath PT, Sharland M. Systematic review of antibiotic resistance rates among Gram-negative bacteria in children with sepsis in resource-limited countries. J Pediatric Infect Dis Soc 2015; 4:11–20 [View Article]
    [Google Scholar]
  6. Saidel-Odes L, Borer A. Limiting and controlling carbapenem-resistant Klebsiella pneumoniae . Infect Drug Resist 2014; 7:9–14
    [Google Scholar]
  7. Gu D, Dong N, Zheng Z, Lin D, Huang M et al. A fatal outbreak of ST11 carbapenem-resistant hypervirulent Klebsiella pneumoniae in a Chinese hospital: a molecular epidemiological study. Lancet Infect Dis 2018; 18:37–46 [View Article]
    [Google Scholar]
  8. Folgori L, Ellis SJ, Bielicki JA, Heath PT, Sharland M et al. Tackling antimicrobial resistance in neonatal sepsis. Lancet Glob Heal 2017; 5:e1066–1068
    [Google Scholar]
  9. Hendlin D, Stapley EO, Jackson M, Wallick H, Miller AK et al. Phosphonomycin, a new antibiotic produced by strains of Streptomyces. Science 1969; 166:122–123 [View Article]
    [Google Scholar]
  10. The World Health Organization Critically important antimicrobials for human medicine, 3rd Revision. 2011
    [Google Scholar]
  11. Pulcini C, Bush K, Craig WA et al. Forgotten antibiotics: an inventory in Europe, the United States, Canada, and Australia. Clin Infect Dis 2012; 54:268–274
    [Google Scholar]
  12. Grabein B, Graninger W, Rodríguez Baño J, Dinh A, Liesenfeld DB. Intravenous fosfomycin—back to the future. Systematic review and meta-analysis of the clinical literature. Clin Microbiol Infect 2017; 23:363–372 [View Article]
    [Google Scholar]
  13. MacLeod DL, Barker LM, Sutherland JL, Moss SC, Gurgel JL et al. Antibacterial activities of a fosfomycin/tobramycin combination: a novel inhaled antibiotic for bronchiectasis. J Antimicrob Chemother 2009; 64:829–836 [View Article]
    [Google Scholar]
  14. Raz R. Fosfomycin: an old—new antibiotic. Clin Microbiol Infect 2012; 18:4–7
    [Google Scholar]
  15. Allerberger F, Klare I. In-vitro activity of fosfomycin against vancomycin-resistant enterococci. J Antimicrob Chemother 1999; 43:211–217
    [Google Scholar]
  16. Falagas ME, Maraki S, Karageorgopoulos DE, Kastoris AC, Mavromanolakis E et al. Antimicrobial susceptibility of multidrug-resistant (MDR) and extensively drug-resistant (XDR) Enterobacteriaceae isolates to fosfomycin. Int J Antimicrob Agents 2010; 35:240–243 [View Article]
    [Google Scholar]
  17. Falagas ME, Kastoris AC, Karageorgopoulos DE, Rafailidis PI. Fosfomycin for the treatment of infections caused by multidrug-resistant non-fermenting Gram-negative bacilli: a systematic review of microbiological, animal and clinical studies. Int J Antimicrob Agents 2009; 34:111–120
    [Google Scholar]
  18. Descourouez JL, Jorgenson MR, Wergin JE, Rose WE. Fosfomycin synergy in vitro with amoxicillin, daptomycin, and linezolid against vancomycin-resistant Enterococcus faecium from renal transplant patients with infected urinary stents. Antimicrob Agents Chemother 2013; 57:1518–1520 [View Article]
    [Google Scholar]
  19. Boyanova L. Susceptibility of anaerobes to fusidic acid and fosfomycin. Int J Antimicrob Agents 2017; 45:560–561
    [Google Scholar]
  20. Frossard M, Joukhadar C, Erovic BM, Dittrich P, Mrass PE et al. Distribution and antimicrobial activity of fosfomycin in the interstitial fluid of human soft tissues. Antimicrob Agents Chemother 2000; 44:2728–2732 [View Article]
    [Google Scholar]
  21. Kuhnen E, Pfeifer G, Frenkel C. Penetration of fosfomycin into cerebrospinal fluid across non-inflamed and inflamed meninges. Infection 1987; 15:422–424
    [Google Scholar]
  22. Sauermann R, Schwameis R, Fille M, Ligios M, Zeitlinger M. Cerebrospinal fluid impairs antimicrobial activity of fosfomycin in vitro . J Antimicrob Chemother 2009; 64:821–823
    [Google Scholar]
  23. Vardakas KZ, Legakis NJ, Triarides N, Falagas ME. Susceptibility of contemporary isolates to fosfomycin: a systematic review of the literature. Int J Antimicrob Agents 2016; 47:269–285
    [Google Scholar]
  24. Garau J. Other antimicrobials of interest in the era of extended-spectrum beta-lactamases: fosfomycin, nitrofurantoin and tigecycline. Clin Microbiol Infect 2008; 14:198–202 [View Article]
    [Google Scholar]
  25. Berkley JA, Lowe BS, Mwangi I et al. Bacteraemia among children admitted to a rural hospital in Kenya. New Engl J Med 2005; 352:39–47
    [Google Scholar]
  26. CLSI Performance Standards for Antimicrobial Susceptibility Testing, 27th ed. CLSI supplement M100. Wayne, PA: Clinical and Laboratory Standards Institute; 2017
    [Google Scholar]
  27. CLSI Performance Standards for Antimicrobial Disk Susceptibility Tests: Approved Standard, 12th ed. CLSI document M02-A12. Wayne, PA: Clinical and Laboratory Standards Institute; 2015
    [Google Scholar]
  28. Greenwood D, Jones A, Eley A. Factors influencing the activity of the trometamol salt of fosfomycin. Eur J Clin Microbiol 1986; 5:29–34
    [Google Scholar]
  29. European Committee on Antimicrobial Susceptibility Testing Fosfomycin: rationale for the EUCAST clinical breakpoints, version 1.0; 2013 http://www.eucast.org/fileadmin/src/media/PDFs/EUCAST_files/Rationale_documents/Fosfomycin_rationale_1.0_20130203.pdf
  30. Walsh CC, McIntosh MP, Peleg AY, Kirkpatrick CM, Bergen PJ. In vitro pharmacodynamics of fosfomycin against clinical isolates of Pseudomonas aeruginosa . J Antimicrob Chemother 2015; 70:3042–3050
    [Google Scholar]
  31. European Committee on Antimicrobial Susceptibility Testing Breakpoint tables for interpretation of MICs and zone diameters, version 2.0; 2012 www.eucast.org
  32. Wachino J, Yamane K, Suzuki S, Kimura K, Arakawa Y. Prevalence of fosfomycin resistance among CTX-M-producing Escherichia coli clinical isolates in Japan and identification of novel plasmid-mediated fosfomycin-modifying enzymes. Antimicrob Agents Chemother 2010; 54:3061–3064
    [Google Scholar]
  33. Folgori L, Livadiotti S, Carletti M, Bielicki J, Pontrelli G et al. Epidemiology and clinical outcomes of multidrug-resistant, gram-negative bloodstream infections in a European tertiary pediatric hospital during a 12-month period. Pediatr Infect Dis J 2014; 33:929–932 [View Article]
    [Google Scholar]
  34. Trecarichi EM, Tumbarello M. Therapeutic options for carbapenem-resistant Enterobacteriaceae infections. Virulence 2017; 8:470–484
    [Google Scholar]
  35. Morrill HJ, Pogue JM, Kaye KS, LaPlante KL. Treatment options for carbapenem-resistant Enterobacteriaceae infections. Open Forum Infect Dis 2015; 2:ofv050
    [Google Scholar]
  36. Endimiani A, Patel G, Hujer KM, Swaminathan M, Perez F et al. In vitro activity of fosfomycin against blaKPC-containing Klebsiella pneumoniae isolates, including those nonsusceptible to tigecycline and/or colistin. Antimicrob Agents Chemother 2010; 54:526–529 [View Article]
    [Google Scholar]
  37. Falagas ME, Maraki S, Karageorgopoulos DE, Kastoris AC, Kapaskelis A et al. Antimicrobial susceptibility of Gram-positive non-urinary isolates to fosfomycin. Int J Antimicrob Agents 2010; 35:497–499 [View Article]
    [Google Scholar]
  38. Falagas ME, Giannopoulou K, Kokolakis G, Rafailidis P. Fosfomycin: use beyond urinary tract and gastrointestinal infections. Clin Infect Dis 2008; 46:1069–1077
    [Google Scholar]
  39. Karageorgopoulos DE, Wang R, Yu XH, Falagas ME. Fosfomycin: evaluation of the published evidence on the emergence of antimicrobial resistance in gram-negative pathogens. J Antimicrob Chemother 2012; 67:255–268 [View Article]
    [Google Scholar]
  40. Jiang Y, Shen P, Wei Z, Liu L, He F et al. Dissemination of a clone carrying a fosA3-harbouring plasmid mediates high fosfomycin resistance rate of KPC-producing Klebsiella pneumoniae in China. Int J Antimicrob Agents 2015; 45:66–70 [View Article]
    [Google Scholar]
  41. European Committee on Antimicrobial Susceptibility Testing Breakpoint tables for interpretation of MICs and zone diameters. http://www.eucast.org/fileadmin/src/media/PDFs/EUCAST_files/Breakpoint_tables/v_7.1_Breakpoint_Tables.pdf
http://instance.metastore.ingenta.com/content/journal/jmm/10.1099/jmm.0.000973
Loading
/content/journal/jmm/10.1099/jmm.0.000973
Loading

Data & Media loading...

This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error